862 research outputs found

    BAYESIAN INTEGRATIVE ANALYSIS OF OMICS DATA

    Get PDF
    Technological innovations have produced large multi-modal datasets that range in multiplatform genomic data, pathway data, proteomic data, imaging data and clinical data. Integrative analysis of such data sets have potentiality in revealing important biological and clinical insights into complex diseases like cancer. This dissertation focuses on Bayesian methodology establishment in integrative analysis of radiogenomics and pathway driver detection applied in cancer applications. We initially present Radio-iBAG that utilizes Bayesian approaches in analyzing radiological imaging and multi-platform genomic data, which we establish a multi-scale Bayesian hierarchical model that simultaneously identifies genomic and radiomic, i.e., radiology-based imaging markers, along with the latent associations between these two modalities, and to detect the overall prognostic relevance of the combined markers. Our method is motivated by and applied to The Cancer Genome Atlas glioblastoma multiforme data set, wherein it identifies important magnetic resonance imaging features and the associated genomic platforms that are also significantly related with patient survival times. For another aspect of integrative analysis, we then present pathDrive that aims to detect key genetic and epigenetic upstream drivers that influence pathway activity. The method is applied into colorectal cancer incorporated with its four molecular subtypes. For each of the pathways that significantly differentiates subgroups, we detect important genomic drivers that can be viewed as “switches” for the pathway activity. To extend the analysis, finally, we develop proteomic based pathway driver analysis for multiple cancer types wherein we simultaneously detect genomic upstream factors that influence a specific pathway for each cancer type within the cancer group. With Bayesian hierarchical model, we detect signals borrowing strength from common cancer type to rare cancer type, and simultaneously estimate their selection similarity. Through simulation study, our method is demonstrated in providing many advantages, including increased power and lower false discovery rates. We then apply the method into the analysis of multiple cancer groups, wherein we detect key genomic upstream drivers with proper biological interpretation. The overall framework and methodologies established in this dissertation illustrate further investigation in the field of integrative analysis of omics data, provide more comprehensive insight into biological mechanisms and processes, cancer development and progression

    The role of network science in glioblastoma

    Get PDF
    Network science has long been recognized as a well-established discipline across many biological domains. In the particular case of cancer genomics, network discovery is challenged by the multitude of available high-dimensional heterogeneous views of data. Glioblastoma (GBM) is an example of such a complex and heterogeneous disease that can be tackled by network science. Identifying the architecture of molecular GBM networks is essential to understanding the information flow and better informing drug development and pre-clinical studies. Here, we review network-based strategies that have been used in the study of GBM, along with the available software implementations for reproducibility and further testing on newly coming datasets. Promising results have been obtained from both bulk and single-cell GBM data, placing network discovery at the forefront of developing a molecularly-informed-based personalized medicine.This work was partially supported by national funds through Fundação para a CiĂȘncia e a Tecnologia (FCT) with references CEECINST/00102/2018, CEECIND/00072/2018 and PD/BDE/143154/2019, UIDB/04516/2020, UIDB/00297/2020, UIDB/50021/2020, UIDB/50022/2020, UIDB/50026/2020, UIDP/50026/2020, NORTE-01-0145-FEDER-000013, and NORTE-01-0145-FEDER000023 and projects PTDC/CCI-BIO/4180/2020 and DSAIPA/DS/0026/2019. This project has received funding from the European Union’s Horizon 2020 research and innovation program under Grant Agreement No. 951970 (OLISSIPO project)

    Integrative pathway enrichment analysis of multivariate omics data

    Full text link
    Multi-omics datasets represent distinct aspects of the central dogma of molecular biology. Such high-dimensional molecular profiles pose challenges to data interpretation and hypothesis generation. ActivePathways is an integrative method that discovers significantly enriched pathways across multiple datasets using statistical data fusion, rationalizes contributing evidence and highlights associated genes. As part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole genome sequencing data from 2658 cancers across 38 tumor types, we integrated genes with coding and non-coding mutations and revealed frequently mutated pathways and additional cancer genes with infrequent mutations. We also analyzed prognostic molecular pathways by integrating genomic and transcriptomic features of 1780 breast cancers and highlighted associations with immune response and anti-apoptotic signaling. Integration of ChIP-seq and RNA-seq data for master regulators of the Hippo pathway across normal human tissues identified processes of tissue regeneration and stem cell regulation. ActivePathways is a versatile method that improves systems-level understanding of cellular organization in health and disease through integration of multiple molecular datasets and pathway annotations

    Pathway-Based Multi-Omics Data Integration for Breast Cancer Diagnosis and Prognosis.

    Get PDF
    Ph.D. Thesis. University of Hawaiʻi at Mānoa 2017

    Statistical Methods in Integrative Genomics

    Get PDF
    Statistical methods in integrative genomics aim to answer important biology questions by jointly analyzing multiple types of genomic data (vertical integration) or aggregating the same type of data across multiple studies (horizontal integration). In this article, we introduce different types of genomic data and data resources, and then review statistical methods of integrative genomics, with emphasis on the motivation and rationale of these methods. We conclude with some summary points and future research directions

    Algorithms for complex systems in the life sciences: AI for gene fusion prioritization and multi-omics data integration

    Get PDF
    Due to the continuous increase in the number and complexity of the genomics and biological data, new computer science techniques are needed to analyse these data and provide valuable insights into the main features. The thesis research topic consists of designing and developing bioinformatics methods for complex systems in life sciences to provide informative models about biological processes. The thesis is divided into two main sub-topics. The first sub-topic concerns machine and deep learning techniques applied to the analysis of aberrant genetic sequences like, for instance, gene fusions. The second one is the development of statistics and deep learning techniques for heterogeneous biological and clinical data integration. Referring to the first sub-topic, a gene fusion is a biological event in which two distinct regions in the DNA create a new fused gene. Gene fusions are a relevant issue in medicine because many gene fusions are involved in cancer, and some of them can even be used as cancer predictors. However, not all of them are necessarily oncogenic. The first part of this thesis is devoted to the automated recognition of oncogenic gene fusions, a very open and challenging problem in cancer development analysis. In this context, an automated model for the recognition of oncogenic gene fusions relying exclusively on the amino acid sequence of the resulting proteins has been developed. The main contributions consist of: 1. creation of a proper database used to train and test the model; 2. development of the methodology through the design and the implementation of a predictive model based on a Convolutional Neural Network (CNN) followed by a bidirectional Long Short Term Memory (LSTM) network; 3. extensive comparative analysis with other reference tools in the literature; 4. engineering of the developed method through the implementation and release of an automated tool for gene fusions prioritization downstream of gene fusion detection tools. Since the previous approach does not consider post-transcriptional regulation effects, new biological features have been considered (e.g., micro RNA data, gene ontologies, and transcription factors) to improve the overall performance, and a new integrated approach based on MLP has explicitly been designed. In the end, extensive comparisons with other methods present in the literature have been made. These contributions led to an improved model that outperforms the previous ones, and it competes with state-of-the-art tools. The rationale behind the second sub-topic of this thesis is the following: due to the widespread of Next Generation Sequencing (NGS) technologies, a large amount of heterogeneous complex data related to several diseases and healthy individuals is now available (e.g., RNA-seq, gene expression data, miRNAs expression data, methylation sequencing data, and many others). Each one of these data is also called omic, and their integrative study is called multi-omics. In this context, the aim is to integrate multi-omics data involving thousands of features (genes, microRNA) and identifying which of them are relevant for a specific biological process. From a computational point of view, finding the best strategies for multi-omics analysis and relevant features identification is a very open challenge. The first chapter dedicated to this second sub-topic focuses on the integrative analysis of gene expression and connectivity data of mouse brains exploiting machine learning techniques. The rational behind this study is the exploration of the capability to evaluate the grade of physical connection between brain regions starting from their gene expression data. Many studies have been performed considering the functional connection of two or more brain areas (which areas are activated in response to a specific stimulus). While, analyzing physical connections (i.e., axon bundles) starting from gene expression data is still an open problem. Despite this study is scientifically very relevant to deepen human brain functioning, ethical reasons strongly limit the availability of samples. For this reason, several studies have been carried out on the mouse brain, anatomically similar to the human one. The neuronal connection data (obtained by viral tracers) of mouse brains were processed to identify brain regions physically connected and then evaluated with these areas’ gene expression data. A multi-layer perceptron was applied to perform the classification task between connected and unconnected regions providing gene expression data as input. Furthermore, a second model was created to infer the degree of connection between distinct brain regions. The implemented models successfully executed the binary classification task (connected regions against unconnected regions) and distinguished the intensity of the connection in low, medium, and high. A second chapter describes a statistical method to reveal pathology-determining microRNA targets in multi-omic datasets. In this work, two multi-omics datasets are used: breast cancer and medulloblastoma datasets. Both the datasets are composed of miRNA, mRNA, and proteomics data related to the same patients. The main computational contribution to the field consists of designing and implementing an algorithm based on the statistical conditional probability to infer the impact of miRNA post-transcriptional regulation on target genes exploiting the protein expression values. The developed methodology allowed a more in-depth understanding and identification of target genes. Also, it proved to be significantly enriched in three well-known databases (miRDB, TargetScan, and miRTarBase), leading to relevant biological insights. Another chapter deals with the classification of multi-omics samples. The literature’s main approaches integrate all the features available for each sample upstream of the classifier (early integration approach) or create separate classifiers for each omic and subsequently define a consensus set rules (late integration approach). In this context, the main contribution consists of introducing the probability concept by creating a model based on Bayesian and MLP networks to achieve a consensus guided by the class label and its probability. This approach has shown how a probabilistic late integration classification is more specific than an early integration approach and can identify samples out of the training domain. To provide new molecular profiles and patients’ categorization, class labels could be helpful. However, they are not always available. Therefore, the need to cluster samples based on their intrinsic characteristics is revealed and dealt with in a specific chapter. Multi-omic clustering in literature is mainly addressed by creating graphs or methods based on multidimensional data reduction. This field’s main contribution is creating a model based on deep learning techniques by implementing an MLP with a specifically designed loss function. The loss represents the input samples in a reduced dimensional space by calculating the intra-cluster and inter-cluster distance at each epoch. This approach reported performances comparable to those of most referred methods in the literature, avoiding pre-processing steps for either feature selection or dimensionality reduction. Moreover, it has no limitations on the number of omics to integrate

    INTEGRATIVE ANALYSIS OF OMICS DATA IN ADULT GLIOMA AND OTHER TCGA CANCERS TO GUIDE PRECISION MEDICINE

    Get PDF
    Transcriptomic profiling and gene expression signatures have been widely applied as effective approaches for enhancing the molecular classification, diagnosis, prognosis or prediction of therapeutic response towards personalized therapy for cancer patients. Thanks to modern genome-wide profiling technology, scientists are able to build engines leveraging massive genomic variations and integrating with clinical data to identify “at risk” individuals for the sake of prevention, diagnosis and therapeutic interventions. In my graduate work for my Ph.D. thesis, I have investigated genomic sequencing data mining to comprehensively characterise molecular classifications and aberrant genomic events associated with clinical prognosis and treatment response, through applying high-dimensional omics genomic data to promote the understanding of gene signatures and somatic molecular alterations contributing to cancer progression and clinical outcomes. Following this motivation, my dissertation has been focused on the following three topics in translational genomics. 1) Characterization of transcriptomic plasticity and its association with the tumor microenvironment in glioblastoma (GBM). I have integrated transcriptomic, genomic, protein and clinical data to increase the accuracy of GBM classification, and identify the association between the GBM mesenchymal subtype and reduced tumorpurity, accompanied with increased presence of tumor-associated microglia. Then I have tackled the sole source of microglial as intrinsic tumor bulk but not their corresponding neurosphere cells through both transcriptional and protein level analysis using a panel of sphere-forming glioma cultures and their parent GBM samples.FurthermoreI have demonstrated my hypothesis through longitudinal analysis of paired primary and recurrent GBM samples that the phenotypic alterations of GBM subtypes are not due to intrinsic proneural-to-mesenchymal transition in tumor cells, rather it is intertwined with increased level of microglia upon disease recurrence. Collectively I have elucidated the critical role of tumor microenvironment (Microglia and macrophages from central nervous system) contributing to the intra-tumor heterogeneity and accurate classification of GBM patients based on transcriptomic profiling, which will not only significantly impact on clinical perspective but also pave the way for preclinical cancer research. 2) Identification of prognostic gene signatures that stratify adult diffuse glioma patientsharboring1p/19q co-deletions. I have compared multiple statistical methods and derived a gene signature significantly associated with survival by applying a machine learning algorithm. Then I have identified inflammatory response and acetylation activity that associated with malignant progression of 1p/19q co-deleted glioma. In addition, I showed this signature translates to other types of adult diffuse glioma, suggesting its universality in the pathobiology of other subset gliomas. My efforts on integrative data analysis of this highly curated data set usingoptimizedstatistical models will reflect the pending update to WHO classification system oftumorsin the central nervous system (CNS). 3) Comprehensive characterization of somatic fusion transcripts in Pan-Cancers. I have identified a panel of novel fusion transcripts across all of TCGA cancer types through transcriptomic profiling. Then I have predicted fusion proteins with kinase activity and hub function of pathway network based on the annotation of genetically mobile domains and functional domain architectures. I have evaluated a panel of in -frame gene fusions as potential driver mutations based on network fusion centrality hypothesis. I have also characterised the emerging complexity of genetic architecture in fusion transcripts through integrating genomic structure and somatic variants and delineating the distinct genomic patterns of fusion events across different cancer types. Overall my exploration of the pathogenetic impact and clinical relevance of candidate gene fusions have provided fundamental insights into the management of a subset of cancer patients by predicting the oncogenic signalling and specific drug targets encoded by these fusion genes. Taken together, the translational genomic research I have conducted during my Ph.D. study will shed new light on precision medicine and contribute to the cancer research community. The novel classification concept, gene signature and fusion transcripts I have identified will address several hotly debated issues in translational genomics, such as complex interactions between tumor bulks and their adjacent microenvironments, prognostic markers for clinical diagnostics and personalized therapy, distinct patterns of genomic structure alterations and oncogenic events in different cancer types, therefore facilitating our understanding of genomic alterations and moving us towards the development of precision medicine
    • 

    corecore